Patients
We recruited a total of 30 male patients with AGA (30–65 years old) according to the 2011 recommendations for the diagnosis and management of gout and hyperuricemia [13], from January 2018 to December 2018 in our hospital. The 30 gouty patients consisted of 17 patients with AGA, which was characterized by the presence of arthritis, and 13 patients with intercritical gout, which was characterized by a confirmed history of gout, but a current absence of symptoms. Exclusion criteria were: failure to consent to the study; suffering from another clinical condition that causes hyperuricemia or cytokine production, such as hemolytic anemia, myeloproliferative disorders, psoriasis, sarcoidosis, acute or chronic renal failure, alcohol intoxication, diabetic ketoacidosis, lactic acidosis, glycogen storage disease type I, hypo- or hyperparathyroidism, or concurrent infections. In addition, 30 age- and gender-matched healthy subjects (27–60 years old) were enrolled as controls. The healthy controls were selected randomly and had no clinical history of gout, arthritis, or rheumatic disease. Blood samples were collected from each participant.
All experimental protocols were approved by the Ethics Committee of Zhejiang Provincial People’s Hospital (Approval number: 2020QT119), and informed consent was obtained from all patients and control subjects.
Whole blood was collected from each subject using heparin anticoagulant tube, centrifuged at 1500 × g for 10 min at 4°C and the plasma thus obtained was frozen at -80°C for the following experiments.
Cell isolation and culture
Neutrophil fraction was separated from fresh peripheral blood using density gradient centrifugation as previously described [14]. Neutrophils were isolated from heparinized blood using commercially available kit (EasySep Human Neutrophil Enrichment Kit, STEMCELL Technologies Inc, Vancouver, Canada). Briefly, to separate polymorphonuclear leukocytes (PMN) from mononuclear cells (PBMC), a standard procedure of Histopaque-1077 (Sigma-Aldrich Chemical Co, St Louis, USA) gradient density centrifugation was used. After centrifugation the layer of plasma and mononuclear cells was discarded and the remaining erythrocytes/granulocytes were transferred into a fresh tube for erythrocytes lysis with a hypotonic ammonium chloride solution.
Obtained PMNs were used for the isolation of neutrophils according to neutrophil enrichment kit protocol provided by the manufacturer.
The isolated neutrophils were maintained in RPMI-1640 medium (Gibco, Carlsbad, CA, USA) supplemented with 10% heat-inactivated fetal bovine serum, 100 U/ml penicillin and 100 µg/ml streptomycin. Neutrophils (1×10
7 cells/ml) were transfected with miR-3146 mimic, inhibitor, or negative controls or stimulated with 1mg/ml monosodium urate (MSU; Sigma-Aldrich, St. Louis, MO, USA) crystals for 2h after treatment with or without 10 mM ROS scavenger N-acetyl-L cystein (NAC; Sigma-Aldrich) for 1h.
In vivo assay
Adult male Sprague-Dawley rats (160-180g), obtained from Center for Animal Experiment of Zhejiang University School of Medicine were used in this study. The rat model of AGA was established via administration of 200µL MSU (20 mg/mL) into the ankle joint cavity. Antagomir-3146 (10 µL) or an equal amount of normal saline was injected at the day of modeling.
The animal procedures were approved by the Institutional Animal Care and Use Committee of Zhejiang University School of Medicine (Approval number: 2019-032).
The synovial tissue specimens were removed from the left knee.
Histopathological examination
The synovial tissues were fixed in 10% paraformaldehyde overnight before decalcification using ethanol and processed for paraffin embedding. Tissue sections (5 µm) were stained with hematoxylin and eosin. Before staining, slides were deparaffinized. To investigate morphologic changes, sections were stained with hematoxylin (Merck, Darmstadt, Germany) and 1% eosin (Sigma-Aldrich), air-dried, and cover-slipped. All slides (× 100 magnification) were observed and photographed using a microscope equipped with camera (BX51; Olympus Ltd., Tokyo, Japan), and images were analyzed using DP2-BSW software (Olympus Ltd).
Measurement of oxidative indicators
The indicators of oxidative damage (MDA, SOD and GSH-Px) were measured using the commercial kits (Beijing Boaosen Biotechnology, Ltd., Beijing, China) following the manufacturer's protocols.
Luciferase reporter assay
HEK-293 cells purchased from the American Type Culture Collection (ATCC, Rockville, MD, USA) were cultured in DMEM supplemented with 10% FBS and 100µg/mL penicillin/streptomycin (all from Gibco, Carlsbad, CA, USA) at 37˚C with 5% CO2.
According to the manufacturer's instructions, luciferase assays were performed in HEK-293 cells after co-transfection with the wild/mutated types of SIRT1 promoter reporters and miR-3146 mimic/mimic NC using Lipofectamine 2000. After 48 h, the cells were collected and tested for luciferase activities using the dual-luciferase reporter assay system (Promega, Madison, WI, USA).
RNA extraction and quantitative real-time PCR
Total RNA was extracted from cells using TRIzol (Invitrogen, Carlsbad, CA, USA) and converted into cDNA using the PrimeScript RT reagent kit (Takara, Dalian, China) according to the manufacturer's instructions. The relative expression levels of miR-3146 and sirtuin 1 (SIRT1) were measured by real time PCR using SYBR Green I real time PCR kit (BD Biosciences, San Diego, CA, USA) on an ABI 7500 Real-Time PCR system (Applied Biosystems, Carlsbad, CA, USA).
The cycling conditions were as follows: initial denaturation at 95˚C for 15 sec, followed by 45 cycles of 55˚C for 33 sec and 70˚C for 30 sec.
The relative expressions were calculated through the 2
−ΔΔCt method. U6 and GAPDH were used as internal controls, respectively. The primers used were as follows: miR-3146, forward: 5'-CATGCTAGGATAGAAAGAATGG-3', reverse: universal primers; SIRT1, forward: 5'-TAGCCTTGTCAGATAAGGAAGGA-3', reverse 5'-ACAGCTTCACAGTCAACTTTGT-3'; U6, forward: 5'-AAAGCAAATCATCGGACGACC-3', reverse 5'-GTACAACACATTGTTTCCTCGGA-3'; GAPDH, forward: 5'-TGTGGGCATCAATGGATTTGG-3', reverse 5'-ACACCATGTATTCCGGGTCAAT-3'.
Western blot
Total proteins were extracted from cultured cells using radio-immunoprecipitation buffer (RIPA). Equal amount of denatured protein samples were separated by electrophoresis and transferred to polyvinylidene fluoride membranes. Immunoblotting was performed at 4°C overnight with appropriate primary antibodies against SIRT1 (#9475; Cell Signaling Technology, Boston, MA, USA) at a dilution of 1:1000. After incubating with HRP-conjugated secondary antibodies (#7074; 1:2000 diltuion, Cell Signaling Technology), the signals were visualized by the ECL system (Thermo Fisher Scientific, Waltham, MA, USA). β-actin was used as a loading control.
Enzyme-linked immuno sorbent assay (ELISA)
According to the manufacturer’s instructions, the quantification of proinflammatory cytokines was conducted by the commercial ELISA kits (Roche Diagnostics, Indianapolis, IN, USA). The optical absorbance was then measured at 405 nm.
IL-8 and IL-1β concentrations in cell culture supernates and serum were quantified using commercial ELISA kits (human IL-8 and IL-1β ELISA reagent kits from R&D Systems, Minneapolis, MN, USA; rat IL-8 and IL-1β ELISA reagent kits from R&D Systems and Shanghai Enzyme-linked Biotechnology Co., Ltd. Shanghai, China), following the manufacturer’s instructions. The microplates were sensitized overnight with coating antibody and blocked with a solution of 4% serine bovine albumin, 5% sucrose in PBS. Supernatants and serum samples and standards were placed in duplicate in the wells of the plate and incubated at 37℃ for 1 h. Samples were washed and detection antibody was added and incubated for 1 h. After appropriate washings, streptavidin conjugated to horseradish peroxidase was added and incubation was performed at 37℃ for 30 min. The reaction was evidenced by the addition of TMB, which was incubated for 10 min at 37℃. The reaction was stopped with H2SO4 and read at 450 nm in an ELISA reader (Microplate Reader, SPECTROstarNano, BGM/LABTECH, Ortemberg, Germany).
For myeloperoxidase (MPO)-DNA complex ELISA assay as previously described [15], a “sandwich” ELISA with anti-MPO monoclonal antibody (Merck Millipore Corp., catalog no. 07–496) and peroxidase-conjugated anti-DNA monoclonal antibody (Roche Diagnostics, Indianapolis, IN, USA; Cell Death Detection ELISA no. 1154467500) was used. Briefly, the wells of microtiter strips were coated with a monoclonal antibody specific for MPO to capture MPO-DNA derived from NETs. A peroxidase substrate was added, which reacted with the bound peroxidase to yield a soluble green product detected at 405 nm. Absorbance readings were proportional to the amount of bound horseradish peroxidase-labeled anti-DNA monoclonal antibody. Results were expressed in arbitrary units.
Immunofluorescence staining
Synovial tissues were digested in Hank's Balanced Salt Solution (Thermo Fisher Scientific, Waltham, MA, USA) containing 0.25% trypsin (Gibco) at 37°C. Treated neutrophils or embedded tissues were fixed in 4% paraformaldehyde for 15 min and permeabilized in 0.01% Triton X-100 for 10 min at room temperature. Samples were then stained with citrullinated histone H3 (CitH3) (1:50 dilution; Abcam, Cambridge, UK) and LYG6 antibodies (1:100 dilution; eBioscience, San Diego, CA, USA). The nucleus was counter-stained with DAPI. Images were captured by confocal microscopy (Zeiss, Jena, Germany) from five randomly fields. The results were expressed as the number of NETs/50 Ly6G-positive cells.
Measurement of intracellular ROS
Intracellular ROS accumulation was measured using dichlorofluorescin diacetate (DCFH-DA; Sigma-Aldrich) as a peroxide-sensitive fluorescent probe at the wavelength 488/525 nm as previously described [16, 17]. Briefly, DCFH-DA was diluted in serum-free DMEM to a final concentration of 10 µM and was added to each well for replacing of original medium. After incubation for 30 min at 37°C, cells were washed thrice with serum-free DMEM. The fluorescence intensities were recorded with a spectrophotometer (Shimadzu Corporation, Tokyo, Japan) with an excitation of 485 nm and an emission of 525 nm.
Statistical analysis
Data from at least three independent experiments were expressed as mean ± standard deviation (SD), analyzed by SPSS 19.0 statistical software (SPSS, Chicago, IL, USA). One-way ANOVA was used to analyze the differences between groups. P < 0.05 indicated a statistically significant difference.